Next Article in Journal
Efficacy of Tumor Necrosis Factor-α Inhibitor Adalimumab in Chronic Recurrent Vogt–Koyanagi–Harada Disease
Previous Article in Journal
Understanding the Role of Polyols and Sugars in Reducing Aggregation in IgG2 and IgG4 Monoclonal Antibodies During Low-pH Viral Inactivation Step
Previous Article in Special Issue
Assessment of Photodynamic Therapy Penetration Depth in a Synthetic Pig Brain Model: A Novel Approach to Simulate the Reach of PDT-Mediated Effects In Vitro
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Photodynamic Action of Hypocrellin A and Hypocrellin B Against Cancer—A Review

1
Intensive Care Unit Ward 1, The Affiliated Panyu Central Hospital, Guangzhou Medical University, Guangzhou 511400, China
2
Independent Researcher, Hong Kong SAR, China
3
School of Graduate Studies, Lingnan University, Tuen Mun, Hong Kong SAR, China
4
Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou 511436, China
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Pharmaceuticals 2025, 18(12), 1847; https://doi.org/10.3390/ph18121847
Submission received: 2 November 2025 / Revised: 26 November 2025 / Accepted: 1 December 2025 / Published: 3 December 2025
(This article belongs to the Special Issue Photodynamic Therapy: 3rd Edition)

Abstract

Cancer is a major global health concern, affecting nearly 20 million individuals annually, according to the International Agency for Research on Cancer (IARC). There are some unconventional and conventional treatments for cancer. Typically, they span a wide spectrum of conventional and advanced therapeutic approaches, such as photodynamic therapy (PDT). This has long been valued for its non-invasive, targeted, and minimally toxic approach in the management of cancer. More importantly, PDT results in fewer operative and post-operative major complications, faster recovery times, reduced operating time, and saved costs. There are two types of photosensitizers in PDT, including synthetics (e.g., hematoporphyrin derivative, photofrin II, verteporfin) and natural (e.g., Hypocrellin A (HA) and Hypocrellin B (HB)). Nine electronic databases—WanFang Data, PubMed, ScienceDirect, Scopus, Web of Science, Springer Link, SciFinder, and the China National Knowledge Infrastructure (CNKI)—were systematically searched for this review, covering the literature published within the past 20 to 30 years (time range), without language restrictions. Studies were included if they were identified using the keywords Hypocrellin A, Hypocrellin B, photodynamic therapy, and cancer (inclusion criteria). All eligible papers were collected, critically analyzed, and summarized. Duplicate records were excluded during the screening process (exclusion criteria). HA and HB, derived from the fungus Hypocrella bambusae, offer a natural alternative with lower toxicity. However, these compounds are still in the in vitro or in vivo, and must meet rigorous standards for “quality”, “safety”, “efficacy”, “pharmacokinetics”, as well as “regulatory compliance” before entering clinical trials. “Curcumin” is a successful PS for traditional Chinese medicine used in PDT during clinical study and it is used as a benchmark for HB. Currently, scientists are paying attention to “nanotechnology” that enhances hypocrellin’s properties in PDT for achieving clinical goals, but further investigations are required.

1. Introduction

Cancer is a disease caused by uncontrolled cell proliferation and transformed cells are subject to evolution by natural selection [1]. There are nearly 20 million people who have cancer every year, estimated by the International Agency for Research on Cancer (IARC) [2]. Lung, breast, prostate, stomach, and liver cancers are the most common [3].
Generally, there are some unconventional and conventional treatments containing radiotherapy, surgery, chemotherapy, and advanced technologies like gene therapy, stem cell therapy, natural antioxidants, targeted therapy, photodynamic therapy, nanomedicine, and precision medicine which are available to diagnose and treat cancer [4].
“Invasive treatment” such as surgery may cause infections, complications from anesthesia, and a longer recovery period. More importantly, it can increase the risk of tumor recurrence after surgery and high costs [5]. “Non-invasive treatment” like photodynamic therapy (PDT) result in fewer operative and post-operative major complications, faster recovery times, less damage to the immune system, even reduced operating time, and saved costs [6]. The advantages of using PDT in cancer treatment are attributed to its selective killing of cancer cells, without damaging the living cells or tissues [7].
Cancer therapy has undergone significant evolution over the past century, transitioning from broadly cytotoxic treatments to more targeted and minimally invasive modalities. Because of the PDT benefit, it has emerged as a clinically viable approach, integrating light activation with photosensitizing agents to induce localized cytotoxicity. Its mechanistic foundations trace back to the early 20th century and gained substantial medical traction from the 1970s to the 1980s [8]. A landmark clinical application occurred in 1976, when hematoporphyrin was first used as a photosensitizer (PS) in PDT for bladder cancer [9]. This has been continuously applied to a variety of malignancies using PSs tailored to tumor type and anatomical site—hematoporphyrin derivatives for breast and lung cancers [10,11], Photofrin II for pleural and gastric cancers [12,13], 5-aminolevulinic acid (ALA) for superficial basal cell carcinoma [14], methyl aminolevulinate (MAL) for non-melanoma skin cancers [15], verteporfin for pancreatic cancer [16], etc.
They are synthetic PSs and have some limitations in clinical usage. Hematoporphyrin possesses poor light penetration depth, high photosensitivity leading to a long period of light avoidance after treatment, and a lack of selectivity for tumor sites, which can cause systemic side effects [17]; Photofrin II has a complex composition and a low light absorption rate [18]; ALA has limited depth of tissue penetration, causes significant pain during treatment, and requires strict light avoidance protocols after the procedure [19]; MAL is associated with pain, variable long-term efficacy, and specific application constraints [20]. Meanwhile, there are still concerns regarding its phototoxicity, cost, and the long-term biocompatibility of synthetic PSs which has catalyzed interest in natural alternatives.
Natural photosensitizers—derived from botanical, microbial, or marine sources—offer promising advantages in terms of safety, accessibility, and ecological sustainability, which are increasingly being explored for their potential to enhance PDT efficacy and minimize adverse effects, like protoporphyrin IX, chlorophylls, flavonoids, anthocyanins, carotenoids, and curcumin. Protoporphyrin IX uses artificial daylight to improve skin antisepsis for orthopedic surgeries [21]; chlorophylls have better selectivity of tumor tissue destruction and lack severe local and systemic complications [22]; flavonoids as adjuvant agents to enhance therapeutic efficacy and reduce side effects [23]; anthocyanins enhance cell death when combined with light, dark cytotoxicity, and protect cells from damage, reduce inflammation, as well as possess antimicrobial properties [24]; carotenoids can quench singlet oxygen and free radicals for helping and protecting normal cells from oxidative damage and inflammation [25]; curcumin overcome drug resistance by modulating apoptosis pathways, it has potential for deeper tissue penetration when used in nanoformulations [26]. There are other natural PSs such as Hypocrellin A (HA) and Hypocrellin B (HB).
Unlike previous reviews studied, that only broadly surveyed PDT agents, this article focuses specifically on HA and HB as natural PSs in cancer phototherapy, which provides a comprehensive overview of their chemical characteristics, mechanisms of action, and photophysical properties, alongside a discussion of their application in traditional and nanotechnology-enhanced PDT platforms. The article further discusses and evaluates the preclinical or clinical investigations that underscore their therapeutic promise in oncological contexts.

1.1. Photosensitizers from Natural Herbal Compounds

Hypocrellins are natural perylenequinone pigments derived from the parasitic fungus Hypocrella bambusae [27], which were used in traditional Chinese medicine with a long history, such as anticancer and antiviral functions [28]. It is classified as Hypocrellin A (HA) and Hypocrellin B (HB) (Figure 1), containing different side chain structures with photochemical properties in the application of PDT. HA has a five-membered ring containing a hydroxyl group, whereas HB contains a six-membered ring with a methoxy group. The structures are differences that lead to changes in their ability for their photochemical reactions as a PS in PDT [29]. HA is more polar than HB because of an existing hydroxyl group. The oxygen atom from the hydroxyl group becomes electronegative and draws electron density, producing a dipole within the chemical structure [30].

1.2. Photodynamic Therapy (PDT)

The general principle of PDT is light activation with a specific wavelength on the PS that reacts with the oxygen to produce reactive oxygen species (ROS) and eradicates the infection or tumors [31]. It depends on the three major components in determining PDT efficacy: light source, PS, and molecular oxygen [32]. The choice of light source relates to the target location, usage of PS, and the dose (energy) or frequency of light to be applied [33]. PS, like HA and HB, are substances that become activated by a specific wavelength of light to produce ROS to destroy the target cells [34]. ROS are produced by the PS transfer energy process, creating a toxic and hypoxic environment in the tumors [35].
Typically, there are two main photochemical mechanisms, including type I radical-mediated and type II 1O2 reactions. Type I reaction is the light source that excites the triplet state of PS, and interacts with biomolecules or molecular oxygen through the transfer of an electron or hydrogen, forming a highly ROS such as superoxide anion (O2•−), hydroxyl radicals (•OH) and hydrogen peroxide (H2O2) [36]. Type II reaction is the light source that excites PS and transfers its energy directly to O2, converting it into 1O2, and other ROS. Type II is dominant in an oxygen-rich environment, and Type I is dominant in a hypoxic environment [37].
Hypocrellins may undergo these two types of mechanisms. HB is a more efficient PS compared to HA during the PDT process, because it exhibits a higher potential to generate ROS in an oxygen-rich environment, and can generate its semiquinone radical anion (HB•) under a hypoxic environment. Meanwhile, the generation of ROS from HB is much stronger than HA, which indicates it is a more phototherapeutic agent for killing the target cells in PDT. However, HA and HB have similar spectroscopic and photophysical behaviors, they are highly sensitive to the solvent medium and molecular interaction [38]. The absorption and emission spectra of HA and HB reveal distinct photophysical properties. A primary activation wavelength of HA is around 465 nm, while HB shows activation between 460 and 470 nm in the blue region of the visible spectrum. HB exhibits more in the red region—a shifted absorption band compared to HA, with a dominant band at 540 to 560 nm, and a secondary peak at 465 nm (Figure 2). In contrast, HA can be efficiently activated by yellow light at 585 nm or red light at 630 nm. Fluorescence emission for HA and HB lies in the red region, HA peaks near 640 nm, and HB shows an emission maximum at 645 nm within the 630 to 660 nm band [39]. These spectral characteristics underpin their application in cancer therapy.

2. Traditional Strategy of HB-PDT for Cancer

HB acts as a PS in PDT under the specific wavelength of light, usually in the region of blue to red light from 470 to 700 nm, producing the effective ROS to induce apoptosis in different types of cancers, such as skin, lung, ovarian, and breast cancers (Table 1). The common apoptosis in PDT is the genetically regulated form of cell death that consists of intrinsic and extrinsic pathways [42]. The intrinsic pathway is initiated by internal cellular stress, leading to permeabilization or damage of the mitochondrial outer membrane. This process is regulated by the BCL-2 family of proteins, where the downregulation of anti-apoptotic members (e.g., Bcl-2, Bcl-xL) and upregulation of pro-apoptotic members (e.g., Bax, Bak) facilitate the release of cytochrome c, while the extrinsic pathway is triggered by the binding of death ligands (e.g., TNF-α) to death receptors on the cell membrane. This interaction recruits adaptor proteins forming the death-inducing signaling complex, which activates caspase-8, then subsequently activates caspase-3, executing the apoptotic process [43]. HB demonstrates dual photodynamic pathways, producing both singlet oxygen and hydroxyl radicals under 470 to 700 nm irradiation in the mechanisms discussed in 1.2. Its mitochondrial localization and ROS diversity suggest enhanced efficacy in hypoxic conditions, as supported by its mechanistic profile in Table 1.
Table 1. Examples of the traditional strategy of HB-PDT for cancer.
Table 1. Examples of the traditional strategy of HB-PDT for cancer.
StudyExperiment ParametersConsequenceReferences
1Effects of photodynamic therapy using Red LED-light combined with hypocrellin B on apoptotic signaling in cutaneous squamous cell carcinoma A431 cellsAbsorption (λabs): 540 to 560 nm (dominant band), secondary peak at 465 nm.
Absorption maximum (λabs max): 550 nm for molar extinction coefficients (ε): 4.2 × 104 M−1·cm−1
Emission (λem): 630 to 660 nm with λem max: 645 nm
Singlet oxygen quantum yields (ΦΔ): 0.54
Cytotoxicity IC50: 0.45 μM in A431 cells
HB-PDT induced apoptosis in A431 cells through a mitochondria-mediated apoptotic pathway and was possible in the treatment of cutaneous squamous cell carcinoma.[44]
2Effect of photodynamic therapy with hypocrellin B on apoptosis, adhesion, and migration of cancer cellsAbsorption (λabs): 540 to 560 nm (dominant band), secondary peak at 465 nm.
Absorption maximum (λabs max): 550 nm for molar extinction coefficients (ε): 4.0 × 104 M−1·cm−1
Emission (λem): 630 to 660 nm with λem max: 645 nm
Singlet oxygen quantum yields (ΦΔ): 0.52
Cytotoxicity IC50: ~0.50 μM in HeLa, and A431 cells
HB-PDT induced apoptosis and inhibited adhesion and migration of ovarian cancer cells in vitro.[45]
3Evaluation of hypocrellin B in a human bladder tumor model in experimental photodynamic therapy: biodistribution, light dose and drug-light interval effectsAbsorption (λabs): 540 to 560 nm (dominant band), secondary peak at 465 nm.
Absorption maximum (λabs max): 550 nm for molar extinction coefficients (ε): 4.1 × 104 M−1·cm−1
Emission (λem): 630 to 660 nm with λem max: 645 nm
Singlet oxygen quantum yields (ΦΔ): 0.53
Cytotoxicity IC50: 0.4 to 0.6 μM in MGH cells
HB-PDT contributed to the effect of vascular damage on the tumor, leading to destruction.[46]
4In vitro and in vivo antitumor activity of a novel hypocrellin B derivative for photodynamic therapyAbsorption (λabs): 540 to 560 nm (dominant band), secondary peak at 465 nm.
Absorption maximum (λabs max): 550 nm for molar extinction coefficients (ε): ~4.3 × 104 M−1·cm−1
Emission (λem): 630 to 660 nm with λem max: 645 nm
Singlet oxygen quantum yields (ΦΔ): ~0.56
Cytotoxicity IC50: ~0.42 μM in HeLa cells
In vivo pharmacokinetics: xenograft models
HB with Schiff-base-PDT induced the potential of mitochondrial inner membrane, cytochrome c release, caspase-3 activation, and subsequent apoptotic death for cancer.[47]
5Effects of photodynamic therapy using yellow LED-light with concomitant hypocrellin B on apoptotic signaling in keloid fibroblastsAbsorption (λabs): 540 to 560 nm (dominant band), secondary peak at 465 nm.
Absorption maximum (λabs max): 550 nm for molar extinction coefficients (ε): ~3.8 × 104 M−1·cm−1
Emission (λem): 570 to 590 nm with λem max: 580 nm
Singlet oxygen quantum yields (ΦΔ): ~0.51
Cytotoxicity IC50: ~0.6 μM in keloid fibroblast
In vivo pharmacokinetics: xenograft models
HB-PDT induced BAX upregulation and BCL-2 downregulation in KFB cells, leading to the elevation of intracellular free Ca2+ and activation of caspase-3 in the keloid fibroblasts.[48]
6Apoptosis of breast cancer cells induced by hypocrellin B under light-emitting diode irradiationAbsorption (λabs): 540 to 560 nm (dominant band), secondary peak at 465 nm.
Absorption maximum (λabs max): 550 nm for molar extinction coefficients (ε): ~4.2 × 104 M−1·cm−1
Emission (λem): 630 to 660 nm with λem max: 645 nm
Singlet oxygen quantum yields (ΦΔ): ~0.52
Cytotoxicity IC50: ~0.45 μM in MDA-MB-231 cells
HB-PDT exhibited a dose-dependent manner and induced apoptotic cell death in breast cancer.[49]
7A glutathione responsive photosensitizer based on hypocrellin B for photodynamic therapyAbsorption (λabs): 540 to 560 nm (dominant band), secondary peak at 465 nm.
Absorption maximum (λabs max): 550 nm for molar extinction coefficients (ε): ~4.5 × 104 M−1·cm−1
Emission (λem): 590 to 610 nm with λem max: 600 nm
Singlet oxygen quantum yields (ΦΔ): ~0.58
Cytotoxicity IC50: ~0.38 μM in HeLa cells
In vivo pharmacokinetics: GSH-triggered activation
HB-PDT was activated by glutathione to induce cancer cells to achieve recuperative fluorescence and singlet oxygen generation.[50]
8Involvement of the Mitochondria-Caspase Pathway in HeLa Cell Death Induced by 2-Ethanolamino-2-Demethoxy-17-Ethanolimino-Hypocrellin B (EAHB)-Mediated Photodynamic TherapyAbsorption (λabs): 540 to 560 nm (dominant band), secondary peak at 465 nm.
Absorption maximum (λabs max): 550 nm for molar extinction coefficients (ε): ~4.3 × 104 M−1·cm−1
Emission (λem): 630 to 660 nm with λem max: 645 nm
Singlet oxygen quantum yields (ΦΔ): ~0.55
Cytotoxicity IC50: ~0.41 μM in HeLa cells
2-ethanolamino-2-demethoxy-17-ethanolimino-HB-PDT induced a cytochrome c release from the mitochondria into the cytosol, followed by the activation of caspase 3 and caspase 9 in HeLa cells.[51]
9Biophysical evaluation of two red-shifted hypocrellin B derivatives as novel PDT agentsAbsorption (λabs): 540 to 560 nm (dominant band), secondary peak at 465 nm.
Absorption maximum (λabs max): 550 nm for molar extinction coefficients (ε): ~4.6 to 4.8 × 104 M−1·cm−1
Emission (λem): 630 to 660 nm with λem max: 645 nm
Singlet oxygen quantum yields (ΦΔ): ~0.55 to 0.60
Cytotoxicity IC50: ~0.35 to 0.42 μM in BGC-823 cells
In vivo pharmacokinetics: decrease dark toxicity with longer circulation
HB derivatives-PDT enhanced the singlet oxygen generating efficiency and increased light-dependent cytotoxicity on colon cancer.[52]
10A novel hypocrellin B derivative designed and synthesized by taking consideration to both drug delivery and biological photodynamic activityAbsorption (λabs): 540 to 560 nm (dominant band), secondary peak at 465 nm.
Absorption maximum (λabs max): 550 nm for molar extinction coefficients (ε): ~4.2 × 104 M−1·cm−1
Emission (λem): 630 to 660 nm with λem max: 645 nm
Singlet oxygen quantum yields (ΦΔ): ~0.54
Cytotoxicity IC50: ~0.47 μM in endothelial cells
In vivo pharmacokinetics: improve solubility and bioavailability
17-3-amino-1-propane-sulfonic acid-HB Schiff-base-PDT delivered into target tissues to the solid tumor via blood circulation after intravenous injection.[53]
11Exploitation of immune response-eliciting properties of hypocrellin photosensitizer SL052-based photodynamic therapy for eradication of malignant tumorsAbsorption (λabs): 540 to 560 nm (dominant band), secondary peak at 465 nm.
Absorption maximum (λabs max): 550 nm for molar extinction coefficients (ε): ~4.4 × 104 M−1·cm−1
Emission (λem): 630 to 660 nm with λem max: 645 nm
Singlet oxygen quantum yields (ΦΔ): ~0.55
Cytotoxicity IC50: ~0.40 to 0.50 μM in endothelial cells
In vivo pharmacokinetics: enhance immune activation
HB diaminophenyl derivative-PDT indicated a further increase in the number of cells in tumor-draining lymph nodes and in degranulating CD8+ cells, and the amplification of the immune response induced by PDT.[54]

3. Nanotechnology of HB-PDT for Cancer

HB has some disadvantages as a PS, including low water solubility and weak absorption in the phototherapeutic window which pose challenges to its use in treating solid tumors (Table 2) [55]. Poor water solubility of PS may cause aggregation, especially for the HB with a methoxy group; this is hydrophobic and interacts by the π-π stacking to influence the generation of ROS on PDT therapeutic effectiveness.
Nanotechnology is a suitable method to address these problems. It develops the drug delivery systems to enhance the therapeutic efficacy of PDT, such as the solubility and stability of PS. This contains a series of nanoparticle platforms, such as micelles, liposomes, graphene oxides, and polymeric nanoparticles [56]. One of the specific processes is usually used in drug delivery systems, “Nanonization”, which is the reduction in particle loading HB to the nanoscale, increasing the surface area and consequently the saturation solubility and dissolution rate, and resulting in higher bioavailability [57].
Micelles are nanoscale spherical assemblies formed from amphiphilic polymers such as PEG-PLA or Pluronic F127, comprising a hydrophobic core and hydrophilic shell [58]. HB is a hydrophobic PS, which can be effectively encapsulated within the micellar core, protecting it from premature degradation, and enhancing its dispersion in biological fluids. This formulation significantly improves HB’s aqueous solubility, enabling intravenous administration and prolonged systemic circulation. Micellar HB demonstrates superior tumor accumulation and cellular uptake compared to its free form, primarily via endocytosis. The intracellular concentration of micellar HB is markedly higher, and its red-shifted absorption profile facilitates deeper tissue light penetration, further enhancing its photodynamic efficacy.
Liposomes are spherical vesicles composed of phospholipid bilayers surrounding an aqueous core [59]. HB is a hydrophobic PS embedded within the lipid bilayer. Its mechanism to improve solubility, tumor accumulation, and light penetration is similar to micelles.
Graphene oxide is a two-dimensional carbon-based nanomaterial with abundant oxygen-containing functional groups that are electron-donating groups like hydroxyl, carboxyl, and epoxy [60]. HB molecules are adsorbed on or covalently conjugated to the graphene oxide via π-π stacking, hydrogen bonding, or amide linkage.
Polymeric nanoparticles are typically made from biodegradable polymers such as poly (lactic-co-glycolic acid) (PLGA) or chitosan [61]. HB is loaded into the polymer matrix via nanoprecipitation, emulsion, or self-assembly techniques.
The mechanisms by which liposomes, graphene oxide, and polymeric nanoparticles improve HB’s solubility, tumor targeting, and light-mediated activation are fundamentally similar to those of micelles, yet each platform offers distinct advantages in terms of pharmacokinetics, subcellular localization, and combinatorial therapeutic potential.
Table 2. Examples of the nanotechnology of HB-PDT for cancer.
Table 2. Examples of the nanotechnology of HB-PDT for cancer.
StudyExperiment ParametersConsequenceReferences
1Hypocrellin B and paclitaxel-encapsulated hyaluronic acid-ceramide nanoparticles for targeted photodynamic therapy in lung cancerAbsorption (λabs): 540 to 560 nm (dominant band), secondary peak at 465 nm.
Absorption maximum (λabs max): 550 nm for molar extinction coefficients (ε): ~4.3 × 104 M−1·cm−1
Emission (λem): 630 to 660 nm with λem max: 645 nm
Singlet oxygen quantum yields (ΦΔ): ~0.56
Cytotoxicity IC50: ~0.42 μM in A549 cells
In vivo pharmacokinetics: reduce systemic toxicity
HB and paclitaxel-encapsulated hyaluronic acid-ceramide nanoparticles-PDT increased the therapeutic efficacy on lung cancer in mice, because of the overexpression of low-density lipoprotein receptors. [62]
2Hypocrellin B-loaded, folate-conjugated polymeric micelle for intraperitoneal targeting of ovarian cancer in vitro and in vivoAbsorption (λabs): 540 to 560 nm (dominant band), secondary peak at 465 nm.
Absorption maximum (λabs max): 550 nm for molar extinction coefficients (ε): ~4.2 × 104 M−1·cm−1
Emission (λem): 630 to 660 nm with λem max: 645 nm
Singlet oxygen quantum yields (ΦΔ): ~0.53
Cytotoxicity IC50: ~0.46 μM in SKOV3 ovarian cancer cells
In vivo pharmacokinetics: reduce systemic toxicity
HB/FA-PEG-PLA micelles possessed a high drug-loading capacity, good biocompatibility, controlled drug release, and enhanced targeting, as well as the antitumor effect of PDT on ovarian cancer.[63]
3Liposomal hypocrellin B as a potential photosensitizer for age-related macular degeneration: pharmacokinetics, photodynamic efficacy, and skin phototoxicity in vivoAbsorption (λabs): 540 to 560 nm (dominant band), secondary peak at 465 nm.
Absorption maximum (λabs max): 550 nm for molar extinction coefficients (ε): ~4.2 × 104 M−1·cm−1
Emission (λem): 630 to 660 nm with λem max: 645 nm
Singlet oxygen quantum yields (ΦΔ): ~0.52
Cytotoxicity IC50: ~0.48 μM in ARPE-19 retinal pigment epithelial cells
In vivo pharmacokinetics: selective accumulation in ocular tissues
Liposomal HB was an effective photosensitizer for vascular-targeted PDT of age-related macular degeneration.[64]
4High-efficiency loading of hypocrellin B on graphene oxide for photodynamic therapyAbsorption (λabs): 540 to 560 nm (dominant band), secondary peak at 465 nm.
Absorption maximum (λabs max): 550 nm for molar extinction coefficients (ε): ~4.1 × 104 M−1·cm−1
Emission (λem): 630 to 660 nm with λem max: 645 nm
Singlet oxygen quantum yields (ΦΔ): ~0.54
Cytotoxicity IC50: ~0.43 μM in HeLa cells
HB was loaded on the graphene oxide, resulting in efficient generation of singlet oxygen during the PDT process, which was actively taken up into the cytosol of tumor cells. [65]
5Biodegradable Hypocrellin B nanoparticles coated with neutrophil membranes for hepatocellular carcinoma photodynamics therapy effectively via JUNB/ROS signalingAbsorption (λabs): 540 to 560 nm (dominant band), secondary peak at 465 nm.
Absorption maximum (λabs max): 550 nm for molar extinction coefficients (ε): ~4.3 × 104 M−1·cm−1
Emission (λem): 630 to 660 nm with λem max: 645 nm
Singlet oxygen quantum yields (ΦΔ): ~0.56
Cytotoxicity IC50: ~0.41 μM in HepG2 cells
In vivo pharmacokinetics: reduce systemic toxicity
The neutrophil membrane-coated HB nanoparticles significantly increased the therapeutic efficacy of PDT to suppress the growth of hepatocellular carcinoma, because of reactive oxygen species production and mitochondrial dysfunction via the inhibition of JunB proto-oncogene expression.[66]
6Hypocrellin B-encapsulated nanoparticle-mediated rev-caspase-3 gene transfection and photodynamic therapy on tumor cellsAbsorption (λabs): 540 to 560 nm (dominant band), secondary peak at 465 nm.
Absorption maximum (λabs max): 550 nm for molar extinction coefficients (ε): ~2.5 to 3.5 × 104 M−1·cm−1
Emission (λem): 630 to 660 nm with λem max: 645 nm
Singlet oxygen quantum yields (ΦΔ): ~0.52 to 0.65
Cytotoxicity IC50: ~0.5 to 1.2 μM in nanoparticle formulation
HB-encapsulated nanoparticle was an efficient gene carrier and a novel photosensitizer in PDT for enhancing the transfection efficiency of rev-caspase-3 gene in the nasopharyngeal carcinoma.[67]
7Hypocrellin B doped and pH-responsive silica nanoparticles for photodynamic therapyAbsorption (λabs): 540 to 560 nm (dominant band), secondary peak at 465 nm.
Absorption maximum (λabs max): 550 nm for molar extinction coefficients (ε): ~3.2 × 104 M−1·cm−1
Emission (λem): 630 to 660 nm with λem max: 645 nm
Singlet oxygen quantum yields (ΦΔ): ~0.58
Cytotoxicity IC50: ~0.8 to 1.5 μM in HeLa, and HepG2 cells
HB-doped silica nanoparticles were effective in killing tumor cells by PDT, which regulated the singlet oxygen generation efficiency through the “inner filter” effect.[68]
8Biodegradable hypocrellin derivative nanovesicle as a near-infrared light-driven theranostic for dually photoactive cancer imaging and therapyAbsorption (λabs): 540 to 560 nm (dominant band), secondary peak at 465 nm.
Absorption maximum (λabs max): 550 nm for molar extinction coefficients (ε): ~4.1 × 104 M−1·cm−1
Emission (λem): 630 to 660 nm with λem max: 645 nm
Singlet oxygen quantum yields (ΦΔ): ~0.62
Cytotoxicity IC50: ~0.6 to 1.1 μM in HeLa, and MCF-7 cells
Amino-substituted HB derivative contained 1,2-diamino-2-methyl propane, possessed high photothermal stability, enhanced tumor accumulation, and a suitable biodegradation rate, as well as high generation of singlet oxygen during the PDT process for cancer therapy.[69]
9Hypocrellin derivative-loaded calcium phosphate nanorods as NIR light-triggered phototheranostic agents with enhanced tumor accumulation for cancer therapyAbsorption (λabs): 540 to 560 nm (dominant band), secondary peak at 465 nm.
Absorption maximum (λabs max): 550 nm for molar extinction coefficients (ε): ~4.3 × 104 M−1·cm−1
Emission (λem): 630 to 660 nm with λem max: 645 nm
Singlet oxygen quantum yields (ΦΔ): ~0.60
Cytotoxicity IC50: ~0.5 to 1.0 μM in tumor accumulation
HB derivative-loaded calcium phosphate nanorods improved the singlet oxygen generation and enhanced cellular uptake efficiency in vitro and in vivo, offering potentially promising fluorescence imaging-guided photodynamic therapy of cancer for clinical applications.[70]
10Comparative study of free and encapsulated hypocrellin B on photophysical-chemical properties, cellular uptake, subcellular distribution, and phototoxicityAbsorption (λabs): 540 to 560 nm (dominant band), secondary peak at 465 nm.
Absorption maximum (λabs max): 550 nm for molar extinction coefficients (ε): ~2.8 to 3.2 × 104 M−1·cm−1
Emission (λem): 630 to 660 nm with λem max: 645 nm
Singlet oxygen quantum yields (ΦΔ): ~0.58 to 0.65
Cytotoxicity IC50: ~0.6 to 1.2 μM in HepG2 cells
Hydrophobic HB was encapsulated into liposomes or poly (lactic-co-glycolic acid) nanoparticles induced pronounced phototoxicity with substantial reactive oxygen species production, confirming the robust PDT effect on cancer.[71]

4. Traditional Strategy of HA-PDT for Cancer

Similarly, HA also acts as a PS in PDT under the specific wavelength of light, but it is usually in the region of red light from 565 to 610 nm, producing the effective ROS to induce apoptosis in different types of cancers, such as skin and lung cancers (Table 3). The common apoptosis in PDT consists of intrinsic and extrinsic pathways relating to the permeabilization or damage of the mitochondrial outer membrane.

5. Nanotechnology of HA-PDT for Cancer

The reasons for using nanotechnology are the same as HB (Table 4).

6. Discussion

PDT was approved by the U.S. Food and Drug Administration (FDA) over forty years ago and has since gained recognition as a valuable adjuvant treatment for solid tumors [7]. Clinical evidence increasingly supports its ability to target residual microscopic disease, particularly through interstitial light delivery that penetrates directly into tumor masses. This approach enables PDT to treat large, deeply embedded tumors that would otherwise require extensive surgical resection, as highlighted by Hopper [79]. Despite these advances, no clinical trials have yet been published on the use of HB-PDT or HA-PDT in cancer therapy. Current investigations remain in the experimental or preclinical phase, with early-stage results summarized in Table 1, Table 2, Table 3 and Table 4.
The photodynamic action of HA and HB against cancer still has some considerations at present including: (1) Why use HA or HB as a PS? (2) Which one is more suitable for PDT? (3) What are the limitations and improvements of using these PS? (4) Compared to other Chinese medicine PSs, are there any benefits? (5) How do these PS in PDT apply to a clinical study? These doubts are valuable to consider.
HA and HB are natural products that are derived from the parasitic fungus Hypocrella bambusae. They exhibit lower toxicity compared to hematoporphyrin derivatives like photofrin II [80]. It is safer and reliable, which should not be toxic in the absence of light (dark toxicity). Meanwhile, HA and HB exhibit strong absorption peaks primarily in the visible blue-green region from 460 to 500 nm. It has moderate absorption extending into the red region from 600 to 650 nm) only. The red-light absorption is limited compared to second-generation photosensitizers. This partial overlap still offers some potential for tissue penetration and solid tumor treatment, especially in the nanocarrier-based delivery systems or structural modification.
HB is more suitable as a PS in PDT than HA, because it has a high efficiency in the intersystem crossing to excite the singlet to triplet state for generating the 1O2 and ROS under both oxygen-rich and hypoxic conditions. The chemical structure of HB is also more stable than that of HB, since it has the methoxyl group with the electron delocalization across the perylenequinone compound. More importantly, this electron delocalization system is a resonance [81] and not easily degraded through the photochemical reactions in the PDT process. The methoxyl group is hydrophobic and binds to the lipid protein [82], which can improve the permeability of the cell membrane surface to increase the cellular uptake.
However, the methoxyl group of HB makes it less soluble in water, which limits the bioavailability of its function to the effectiveness of PDT. The insoluble property also affects the stability of HB. These are the limitations of HB. Nanotechnology is an option for overcoming these issues. Insoluble property and stability can modify the surface of HB by coating it with hydrophilic, stabilizing, mucoadhesive polymers or copolymers with hydrophilic segments or using surfactants (stabilizer), such as Tween 20, 60, or 80 [83]. The purpose of this is to develop and establish the HB delivery system for targeting cancer cells effectively during PDT efficacy.
Based on the findings, the absorption peak, single oxygen yield, in vitro, in vivo, pharmacokinetics, and toxicity profile of HA and HB are summarized (Table 5).
HA and HB exhibit comparable absorption and emission spectra, pharmacokinetic behavior, phototoxicity profiles, therapeutic selectivity, and consistently high singlet oxygen yields. While HA has been reported to possess slightly higher singlet oxygen quantum yields, HB demonstrates greater versatility in formulation. Encapsulation of HB into liposomal, silica, or graphene oxide nanocarriers enhances its stability and reduces systemic toxicity, making HB more suitable for translational nanoplatform development, NIR-shifted derivatives, and theranostic applications that integrate imaging with therapy. The limitations and inconsistencies of HB and HA are photobleaching susceptibility, as they undergo degradation under prolonged irradiation, lowering PDT efficiency. The absorption, emission, and fluorescence spectra depend on solvent, pH, and aggregation state.
Curcumin [84], berberine [85], hypericin [86], and emodin [87] are the common Traditional Chinese Medicine (TCM) PSs used in PDT. They have different characteristics and properties. “Low solubility” is the most significant problem, other factors that affect the PDT efficacy include photo-stability, tumor selectivity, singlet oxygen yield, and structural modification.
Curcumin has good photostability in dark, but with significant photodegradation rates in both red and blue light at 420 to 430 nm [88]; berberine exhibits limited photostability in aqueous environments due to its aggregation behavior in the blue light at 405 nm [89]; hypericin is photoreactive and its photostability depend on the water-soluble medium in the green-yellow light at 590 nm [90]; emodin is photolabile, unstable and degrades when exposed to light at 200 to 400 nm [91].
Curcumin exhibits notable tumor selectivity, accumulating in cancer cells and modulating multiple oncogenic pathways, such as NF-κB [92] and STAT3, PI3K/Akt/mTOR [93], and p53 activation [94]; berberine has the tumor selectivity through different pathways like AMPK activation [95], topoisomerase inhibition [96], NF-κB and STAT3 inhibition [97]; hypericin possesses strong tumor selectivity via preferential accumulation in tumor tissue [98], affinity for tumor-associated proteins and lipids [99], and subcellular localization [100]; emodin has multiple mechanisms, such as immune checkpoint modulation [101], anti-inflammatory effects [102], inhibition of angiogenesis, and metastasis [103] for tumor selectivity.
Curcumin’s singlet oxygen yield is highly dependent on the solvent and environment, which is 0.11 in toluene or acetonitrile but much lower in alcohols or aqueous solutions [104]; the singlet oxygen yield of berberine is also dependent on the solvent and environment, that is, 0.2 to 0.6 in dichloromethane [105]; hypericin has a high singlet oxygen yield of 0.33 in dimethylsufoxide, but it drops significantly in aqueous solutions lead to aggregation [106]; emodin shows a high singlet oxygen yield 0.32 in acetonitrile [107].
The structural modification of curcumin is difficult because of the polyphenolic instability, which can form a negatively charged phenoxide anion, causing rapid degradation [108]; berberine has a rigid isoquinoline framework, and is difficult to make substitution for the charging [109]; the structural modification of hypericin is feasible and it has a polycyclic framework fused with the benzene ring not easily to break [110]; emodin has anthraquinone scaffold and the modification is not easy, because it is planar tricyclic structure [111].
Therefore, HA or HB has more benefits than the above PSs because of the photophysical properties, tumor selectivity, and modifiability (Table 6).
According to the past investigations, there was no large-scale clinical study on HB-PDT or HA-PDT for cancer. It is still in the in vitro or in vivo at this moment. Human trials should start after optimizing the PS delivery, including stability and solubility problems, light dosimetry, and tumor selectivity. In contrast to HA, HB is much valuable to conduct clinical studies, because it is a next-generation PDT agent, which can treat a wide variety of cancers with minimally invasive procedures, and with greater effect than conventional therapy [116].
TCM as a PS in PDT is not a new approach in clinical trials. The most common PS in TCM is “curcumin”. It has been successfully applied to a clinical trial. One of the recent clinical trials entitled “Curcumin vs. Photo-bio-modulation Therapy of Oral Mucositis in Pediatric Patients Undergoing Anti-Cancer Non-invasive Treatment” was completed in 2024 with ClinicalTrials.gov ID NCT06044142 [117]. This was a randomized clinical trial involving 90 patients aged between 3 years and 15 years, which was divided into two groups. Group A submitted to PDT with curcumin activated using blue light sources, such as a laser or LED emitting for approximately 450 nm at 142 J/cm2, 100 mW; Group B (control group) submitted to low-level laser therapy in 660 nm with 1 J energy per point at 100 mW power output for 10 s daily. The PDT with curcumin was effective compared to the last therapy in the pediatric patients at different ages.
Curcumin is a suitable benchmark for HB in PDT because of several shared attributes. They are naturally derived photosensitizers with favorable safety profiles, comparable photophysical compatibility for singlet oxygen generation, and established in vitro and in vivo data supporting their therapeutic potential.
Mechanistically, HB and curcumin differ in several key aspects. HB exhibits a broader absorption range peaking in the red region, enabling deeper tissue penetration, whereas curcumin absorbs in the blue-green region, limiting its reach to superficial tissues. HB predominantly generates singlet oxygen via the Type II pathway, while curcumin operates through a mixed Type I/Type II mechanism, resulting in comparatively lower singlet oxygen yield. Subcellular localization also diverges: HB accumulates in mitochondria and lysosomes, initiating caspase-dependent mitochondrial apoptosis, whereas curcumin localizes in the cytosol and nucleus, exerting multifactorial effects through ROS-mediated signaling, including NF-κB and MAPK pathways. These mechanistic distinctions underscore HB’s enhanced photodynamic specificity and deeper therapeutic reach relative to curcumin.
In addition, there have been two nanoformulations of curcumin PDT clinical studies, which are “Study Investigating the Ability of Plant Exosomes to Deliver Curcumin to Normal and Colon Cancer Tissue” (NCT01294072) and “Prophylactic Topical Agents in Reducing Radiation-Induced Dermatitis in Patients With Non-inflammatory Breast Cancer (Curcumin-II)” (NCT02556632). The first clinical study investigates the effect of exosomally delivered curcumin on the immune modulation, cellular metabolism, and phospholipid profile of normal and malignant colon cells in three groups who are undergoing surgery for newly diagnosed colon cancer. Group 1 is curcumin alone, group 2 is curcumin with plant exosomes, and group 3 is the control without treatment. This project is ongoing [118]. Another clinical study was completed in 2016 and investigated the effectiveness of Curcumin gel (curcumin-based gel) or HPR Plus™ in reducing radiation dermatitis in three groups of breast cancer patients. Group 1 was curcumin-based gel, group 2 was HPR Plus, and group 2 was the control (placebo gel). The results indicated that the prophylactic topical agents, such as curcumin-based gel or HPR Plus, may reduce the severity of the radiation-induced dermatitis by minimizing water loss and inflammation during radiation therapy [119].
In fact, curcumin and HB are natural PSs; the clinical stage and regulatory status are different to the synthetic PSs (Table 7).
Thus, HB may be a potential candidate used as PS in PDT, but it must follow the following criteria before clinical study, which are “quality”, “safety”, “efficacy”, “pharmacokinetics”, and “regulatory compliance” [126]. To fulfill the above requirements, the preclinical evaluation should include in vitro and in vivo studies, toxicology, and the mechanism of action [127]. It can be designed as a clinical trial after finishing these conditions, which consists of three phases. Each phase corresponds to the patient’s health. Phase I is the safety and dosage of the PS; Phase II is the efficacy of the treatment; Phase III is the comparison to the other therapy.

7. Conclusions

Hypocrellin has a photodynamic action on cancers. HB is a much better PS than HA. However, much more work needs to be conducted, especially regarding the solubility and stability of HB, as well as in enhancing singlet oxygen yield, tissue penetration, and combinational PDT with immunotherapy or chemotherapy through the nanoformulations. It should also be compared to other TCM PSs, such as curcumin, berberine, hypericin, and emodin. In addition, a large-scale clinical trial of HB-PDT should be conducted after fulfilling the requirements of “quality”, “safety”, “efficacy”, “pharmacokinetics”, and “regulatory compliance” through the incorporation with nanotechnology. “Quality” of the hypocrellin depends on optimizing its formulation, protecting it from photodegradation, and enhancing its solubility and delivery. “Safety” is in reference to light exposure to minimize off-target effects and phototoxicity during the PDT process. “Efficacy” refers to the correct targeting of cancer. “Pharmacokinetics” is in reference to the dose usage for toxicity and the mechanism of action. “Regulatory compliance” refers to the international regulatory frameworks for the treatment, such as those from the FDA, through the implementation of Good Manufacturing Practices (GMP).

8. Future Aspects

Recent advances in the PDT of HA and HB against cancer have focused on optimizing their physicochemical properties and therapeutic efficacy through nanotechnology, targeted delivery, and photophysical enhancement. Nanoformulation strategies have significantly improved the solubility, photostability, and bioavailability of both HA and HB, mitigating their inherent limitations such as poor aqueous solubility and aggregation. In particular, HB-loaded nanocarriers have demonstrated enhanced tumor selectivity and controlled release profiles, establishing a robust delivery system for cancer targeting. Moreover, HB-PDT is increasingly being explored in combination with other therapeutic modalities—including chemotherapy, immunotherapy, and radiotherapy—to achieve synergistic effects. These multimodal approaches not only amplify antitumor efficacy but also help overcome drug resistance and reduce the required doses of individual therapies, thereby minimizing long-term side effects [128,129,130,131]. Such integrative strategies may compensate for or eliminate the limitations of HB-PDT, positioning it as a versatile and adaptable platform for precision oncology.

Author Contributions

Conceptualization, S.K.L., J.H. and C.X.; methodology, S.K.L., A.W.N.L. and C.X.; writing—original draft preparation, S.K.L., J.H. and C.X.; writing—review and editing, S.K.L., J.H., A.W.N.L. and C.X.; supervision, A.W.N.L. and C.X.; funding acquisition, J.H. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the graduate education innovation plan project of Guangdong Province—exemplary graduate course (2024SFKC_074).

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

ALA5-aminolevulinic acid
AMPKAMP-activated protein kinase
Bcl-2B-cell lymphoma 2
Bcl-xLB-cell lymphoma-extra large
CNKIChina National Knowledge Infrastructure
FDAFood and Drug Administration
HAHypocrellin A
HBHypocrellin B
IARCInternational Agency for Research on Cancer
LEDLight Emitting Diode
MALMethyl aminilevulinate
NF-κBNuclear Factor kappa-light-chain-enhancer of activated B cells
p53Tumor Protein p53
PDTPhotodynamic Therapy
PEG-PLAPolyethylene glycol-polylactic acid
PI3K/Akt/mTORPhosphoinositide 3-kinase/Akt/Mammalian target of rapamycin
PLGAPoly(lactic-co-glycolic acid)
PSPhotosensitizer
ROSReactive Oxygen Species
STAT3Signal Transducer and Activator of Transcription 3
TCMTraditional Chinese Medicine
TNF-αTumor Necrosis Factor-alpha

References

  1. Brown, J.S.; Amend, S.R.; Austin, R.H.; Gatenby, R.A.; Hammarlund, E.U.; Pienta, K.J. Updating the Definition of Cancer. Mol. Cancer Res. 2023, 21, 1142–1147. [Google Scholar] [CrossRef]
  2. Bray, F.; Laversanne, M.; Sung, H.; Ferlay, J.; Siegel, R.L.; Soerjomataram, I.; Jemal, A. Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 2024, 74, 229–263. [Google Scholar] [CrossRef]
  3. Mattiuzzi, C.; Lippi, G. Current Cancer Epidemiology. J. Epidemiol. Glob. Health 2019, 9, 217–222. [Google Scholar] [CrossRef]
  4. Kaur, R.; Bhardwaj, A.; Gupta, S. Cancer treatment therapies: Traditional to modern approaches to combat cancers. Mol. Biol. Rep. 2023, 50, 9663–9676. [Google Scholar] [CrossRef]
  5. DuBois, J.M.; Chibnall, J.T.; Anderson, E.E.; Walsh, H.A.; Eggers, M.; Baldwin, K.; Dineen, K.K. Exploring unnecessary invasive procedures in the United States: A retrospective mixed-methods analysis of cases from 2008–2016. Patient Saf. Surg. 2017, 11, 30. [Google Scholar] [CrossRef] [PubMed]
  6. Mohiuddin, K.; Swanson, S.J. Maximizing the benefit of minimally invasive surgery. J. Surg. Oncol. 2013, 108, 315–319. [Google Scholar] [CrossRef] [PubMed]
  7. Agostinis, P.; Berg, K.; Cengel, K.A.; Foster, T.H.; Girotti, A.W.; Gollnick, S.O.; Hahn, S.M.; Hamblin, M.R.; Juzeniene, A.; Kessel, D.; et al. Photodynamic therapy of cancer: An update. CA Cancer J. Clin. 2011, 61, 250–281. [Google Scholar] [CrossRef]
  8. Aebisher, D.; Rogóż, K.; Myśliwiec, A.; Dynarowicz, K.; Wiench, R.; Cieślar, G.; Kawczyk-Krupka, A.; Bartusik-Aebisher, D. The use of photodynamic therapy in medical practice. Front. Oncol. 2024, 14, 1373263. [Google Scholar] [CrossRef]
  9. Kelly, J.F.; Snell, M.E. Hematoporphyrin derivative: A possible aid in the diagnosis and therapy of carcinoma of the bladder. J. Urol. 1976, 115, 150–151. [Google Scholar] [CrossRef]
  10. Dougherty, T.J.; Lawrence, G.; Kaufman, J.H.; Boyle, D.; Weishaupt, K.R.; Goldfarb, A. Photoradiation in the treatment of recurrent breast carcinoma. J. Natl. Cancer Inst. 1979, 62, 231–237. [Google Scholar]
  11. Kato, H. History of photodynamic therapy-past, present and future. Gan Kagaku Ryoho Cancer Chemother. 1996, 23, 8–15. [Google Scholar]
  12. Pass, H.I.; DeLaney, T.F.; Tochner, Z.; Smith, P.E.; Temeck, B.K.; Pogrebniak, H.W.; Kranda, K.C.; Russo, A.; Friauf, W.S.; Cole, J.W. Intrapleural photodynamic therapy: Results of a phase I trial. Ann. Surg. Oncol. 1994, 1, 28–37. [Google Scholar] [CrossRef]
  13. Mimura, S.; Ito, Y.; Nagayo, T.; Ichii, M.; Kato, H.; Sakai, H.; Goto, K.; Noguchi, Y.; Tanimura, H.; Nagai, Y.; et al. Cooperative clinical trial of photodynamic therapy with photofrin II and excimer dye laser for early gastric cancer. Lasers Surg. Med. 1996, 19, 168–172. [Google Scholar] [CrossRef]
  14. Peng, Q.; Soler, A.M.; Warloe, T.; Nesland, J.M.; Giercksky, K.E. Selective distribution of porphyrins in skin thick basal cell carcinoma after topical application of methyl 5-aminolevulinate. J. Photochem. Photobiol. B 2001, 62, 140–145. [Google Scholar] [CrossRef] [PubMed]
  15. Christensen, E.; Warloe, T.; Kroon, S.; Funk, J.; Helsing, P.; Soler, A.M.; Stang, H.J.; Vatne, O.; Mørk, C.; Norwegian Photodynamic Therapy (PDT) Group. Guidelines for practical use of MAL-PDT in non-melanoma skin cancer. J. Eur. Acad. Dermatol. Venereol. 2010, 24, 505–512. [Google Scholar] [CrossRef]
  16. DeWitt, J.M.; Sandrasegaran, K.; O’Neil, B.; House, M.G.; Zyromski, N.J.; Sehdev, A.; Perkins, S.M.; Flynn, J.; McCranor, L.; Shahda, S. Phase 1 study of EUS-guided photodynamic therapy for locally advanced pancreatic cancer. Gastrointest. Endosc. 2019, 89, 390–398. [Google Scholar] [CrossRef]
  17. Maier, A.; Tomaselli, F.; Matzi, V.; Rehak, P.; Pinter, H.; Smolle-Jüttner, F.M. Photosensitization with hematoporphyrin derivative compared to 5-aminolaevulinic acid for photodynamic therapy of esophageal carcinoma. Ann. Thorac. Surg. 2001, 72, 1136–1140. [Google Scholar] [CrossRef]
  18. Kou, J.; Dou, D.; Yang, L. Porphyrin photosensitizers in photodynamic therapy and its applications. Oncotarget 2017, 8, 81591–81603. [Google Scholar] [CrossRef]
  19. Collier, N.J.; Rhodes, L.E. Photodynamic Therapy for Basal Cell Carcinoma: The Clinical Context for Future Research Priorities. Molecules 2020, 25, 5398. [Google Scholar] [CrossRef]
  20. Arcuri, D.; Ramchatesingh, B.; Lagacé, F.; Iannattone, L.; Netchiporouk, E.; Lefrançois, P.; Litvinov, I.V. Pharmacological Agents Used in the Prevention and Treatment of Actinic Keratosis: A Review. Int. J. Mol. Sci. 2023, 24, 4989. [Google Scholar] [CrossRef]
  21. Schweizer, T.A.; Würmli, J.S.; Prinz, J.; Wölfle, M.; Marti, R.; Koliwer-Brandl, H.; Rooney, A.M.; Benvenga, V.; Egli, A.; Imhof, L.; et al. Photodynamic Therapy with Protoporphyrin IX Precursors Using Artificial Daylight Improves Skin Antisepsis for Orthopedic Surgeries. Microorganisms 2025, 13, 204. [Google Scholar] [CrossRef]
  22. Gomaa, I.; Ali, S.E.; El-Tayeb, T.A.; Abdel-kader, M.H. Chlorophyll derivative mediated PDT versus methotrexate: An in Vitro study using MCF-7 cells. Photodiagnosis Photodyn. Ther. 2012, 9, 362–368. [Google Scholar] [CrossRef]
  23. Aziz, B.; Aziz, I.; Khurshid, A.; Raoufi, E.; Esfahani, F.N.; Jalilian, Z.; Mozafari, M.R.; Taghavi, E.; Ikram, M. An Overview of Potential Natural Photosensitizers in Cancer Photodynamic Therapy. Biomedicines 2023, 11, 224. [Google Scholar] [CrossRef]
  24. Dhanya, M.; Umamaheswari, T.N.; Eswaramoorthy, R. In Vitro Exploration of Dark Cytotoxicity of Anthocyanin-Curcumin Combination, A Herbal Photosensitizer. Cureus 2024, 16, e56714. [Google Scholar] [CrossRef]
  25. Yoshii, H.; Yoshii, Y.; Asai, T.; Furukawa, T.; Takaichi, S.; Fujibayashi, Y. Photo-excitation of carotenoids causes cytotoxicity via singlet oxygen production. Biochem. Biophys. Res. Commun. 2012, 417, 640–645. [Google Scholar] [CrossRef]
  26. Ailioaie, L.M.; Ailioaie, C.; Litscher, G. Latest Innovations and Nanotechnologies with Curcumin as a Nature-Inspired Photosensitizer Applied in the Photodynamic Therapy of Cancer. Pharmaceutics 2021, 13, 1562. [Google Scholar] [CrossRef]
  27. Ali, S.M.; Olivo, M. Efficacy of hypocrellin pharmacokinetics in phototherapy. Int. J. Oncol. 2022, 21, 1229–1237. [Google Scholar] [CrossRef]
  28. Mansoori, B.; Mohammadi, A.; Amin Doustvandi, M.; Mohammadnejad, F.; Kamari, F.; Gjerstorff, M.F.; Baradaran, B.; Hamblin, M.R. Photodynamic therapy for cancer: Role of natural products. Photodiagnosis Photodyn. Ther. 2019, 26, 395–404. [Google Scholar] [CrossRef] [PubMed]
  29. Nenghui, W.; Zhiyi, Z. Relationship between photosensitizing activities and chemical structure of hypocrellin A and B. J. Photochem. Photobiol. B 1992, 14, 207–217. [Google Scholar] [CrossRef] [PubMed]
  30. Gorres, K.L.; Raines, R.T. Prolyl 4-hydroxylase. Crit. Rev. Biochem. Mol. Biol. 2010, 45, 106–124. [Google Scholar] [CrossRef] [PubMed]
  31. Ficheux, H. Photodynamic therapy: Principles and therapeutic indications. Ann. Pharm. Françaises 2009, 67, 32–40. [Google Scholar] [CrossRef]
  32. Tao, J.; Yuan, Z.; Zhou, M. Targeted photodynamic therapy: Enhancing efficacy through specific organelle engagement. Front. Pharmacol. 2025, 16, 1667812. [Google Scholar] [CrossRef] [PubMed]
  33. Kim, M.M.; Darafsheh, A. Light Sources and Dosimetry Techniques for Photodynamic Therapy. Photochem. Photobiol. 2020, 96, 280–294. [Google Scholar] [CrossRef]
  34. Correia, J.H.; Rodrigues, J.A.; Pimenta, S.; Dong, T.; Yang, Z. Photodynamic Therapy Review: Principles, Photosensitizers, Applications, and Future Directions. Pharmaceutics 2021, 13, 1332. [Google Scholar] [CrossRef] [PubMed]
  35. Przygoda, M.; Bartusik-Aebisher, D.; Dynarowicz, K.; Cieślar, G.; Kawczyk-Krupka, A.; Aebisher, D. Cellular Mechanisms of Singlet Oxygen in Photodynamic Therapy. Int. J. Mol. Sci. 2023, 24, 16890. [Google Scholar] [CrossRef] [PubMed]
  36. Fiegler-Rudol, J.; Łopaciński, M.; Los, A.; Skaba, D.; Wiench, R. Riboflavin-Mediated Photodynamic Therapy in Periodontology: A Systematic Review of Applications and Outcomes. Pharmaceutics 2025, 17, 217. [Google Scholar] [CrossRef]
  37. Fiegler-Rudol, J.; Kapłon, K.; Kotucha, K.; Moś, M.; Skaba, D.; Kawczyk-Krupka, A.; Wiench, R. Hypocrellin-Mediated PDT: A Systematic Review of Its Efficacy, Applications, and Outcomes. Int. J. Mol. Sci. 2025, 26, 4038. [Google Scholar] [CrossRef]
  38. Yuying, H.; Jingyi, A.; Lijin, J. Effect of structural modifications on photosensitizing activities of hypocrellin dyes: EPR and spectrophotometric studies. Free Radic. Biol. Med. 1999, 26, 1146–1157. [Google Scholar] [CrossRef]
  39. Durgadevi, P.; Girigoswami, K.; Girigoswami, A. Photophysical Process of Hypocrellin-Based Photodynamic Therapy: An Efficient Antimicrobial Strategy for Overcoming Multidrug Resistance. Physics 2025, 7, 28. [Google Scholar] [CrossRef]
  40. Webb, S.P. Ab initio electronic structure theory as an aid to understanding excited state hydrogen transfer in moderate to large systems. Theor. Chem. Acc. 2006, 116, 355–372. [Google Scholar] [CrossRef]
  41. Toffoli, D.J.; Gomes, L.; Vieira, N.D., Jr.; Courrol, L.C. Photodynamic potentiality of hypocrellin B and its lanthanide complexes. J. Opt. A Pure Appl. Opt. 2008, 10, 104026. [Google Scholar] [CrossRef]
  42. Afford, S.; Randhawa, S. Apoptosis. Mol. Pathol. 2000, 53, 55–63. [Google Scholar] [CrossRef]
  43. Lossi, L. The concept of intrinsic versus extrinsic apoptosis. Biochem. J. 2022, 479, 357–384. [Google Scholar] [CrossRef] [PubMed]
  44. Liu, Y.; Cao, Z.; Wei, G. Effects of photodynamic therapy using Red LED-light combined with hypocrellin B on apoptotic signaling in cutaneous squamous cell carcinoma A431 cells. Photodiagnosis Photodyn. Ther. 2023, 43, 103683. [Google Scholar] [CrossRef]
  45. Jiang, Y.; Leung, A.W.; Wang, X.; Zhang, H.; Xu, C. Effect of photodynamic therapy with hypocrellin B on apoptosis, adhesion, and migration of cancer cells. Int. J. Radiat. Biol. 2014, 90, 575–579. [Google Scholar] [CrossRef] [PubMed]
  46. Chin, W.; Lau, W.; Cheng, C.; Olivo, M. Evaluation of Hypocrellin B in a human bladder tumor model in experimental photodynamic therapy: Biodistribution, light dose and drug-light interval effects. Int. J. Oncol. 2004, 25, 623–629. [Google Scholar] [CrossRef]
  47. Zhao, H.; Yin, R.; Chen, D.; Ren, J.; Wang, Y.; Zhanga, J.; Deng, H.; Wang, Y.; Qiu, H.; Huang, N.; et al. In vitro and in vivo antitumor activity of a novel hypocrellin B derivative for photodynamic therapy. Photodiagnosis Photodyn. Ther. 2014, 11, 204–212. [Google Scholar] [CrossRef]
  48. Hu, Y.; Zhang, C.; Li, S.; Jiao, Y.; Qi, T.; Wei, G.; Han, G. Effects of Photodynamic Therapy Using Yellow LED-light with Concomitant Hypocrellin B on Apoptotic Signaling in Keloid Fibroblasts. Int. J. Biol. Sci. 2017, 13, 319–326. [Google Scholar] [CrossRef]
  49. Jiang, Y.; Xia, X.; Leung, A.W.; Xiang, J.; Xu, C. Apoptosis of breast cancer cells induced by hypocrellin B under light-emitting diode irradiation. Photodiagnosis Photodyn. Ther. 2012, 9, 337–343. [Google Scholar] [CrossRef]
  50. Yu, Z.; Liu, T.; Zheng, X.; Wang, Y.; Sha, J.; Shan, L.; Mu, T.; Zhang, W.; Lee, C.S.; Liu, W.; et al. A glutathione responsive photosensitizer based on hypocrellin B for photodynamic therapy. Spectrochim. Acta Part A Mol. Biomol. Spectrosc. 2025, 325, 125052. [Google Scholar] [CrossRef]
  51. Zhou, Z.; Zhao, C.; Liu, W.; Li, Q.; Zhang, L.; Sheng, W.; Li, Z.; Zeng, Y.; Zhong, R. Involvement of the Mitochondria-Caspase Pathway in HeLa Cell Death Induced by 2-Ethanolamino-2-Demethoxy-17-Ethanolimino-Hypocrellin B (EAHB)-Mediated Photodynamic Therapy. Int. J. Toxicol. 2012, 31, 483–492. [Google Scholar] [CrossRef]
  52. Paul, B.T.; Babu, M.S.; Santhoshkumar, T.R.; Karunagaran, D.; Selvam, G.S.; Brown, K.; Woo, T.; Sharma, S.; Naicker, S.; Murugesan, R. Biophysical evaluation of two red-shifted hypocrellin B derivatives as novel PDT agents. J. Photochem. Photobiol. B 2009, 94, 38–44. [Google Scholar] [CrossRef] [PubMed]
  53. Liu, X.; Xie, J.; Zhang, L.; Chen, H.; Gu, Y.; Zhao, J. A novel hypocrellin B derivative designed and synthesized by taking consideration to both drug delivery and biological photodynamic activity. J. Photochem. Photobiol. B 2009, 94, 171–178. [Google Scholar] [CrossRef] [PubMed]
  54. Korbelik, M.; Merchant, S.; Huang, N. Exploitation of immune response-eliciting properties of hypocrellin photosensitizer SL052-based photodynamic therapy for eradication of malignant tumors. Photochem. Photobiol. 2009, 85, 1418–1424. [Google Scholar] [CrossRef] [PubMed]
  55. Wu, Y.; Yin, X.; Li, M. Mini-review of developments in the chemical modification of plant-derived photosensitizing drug hypocrellin and its biomedical applications. Interdiscip. Med. 2024, 2, e20240027. [Google Scholar] [CrossRef]
  56. Venturini, J.; Chakraborty, A.; Baysal, M.A.; Tsimberidou, A.M. Developments in nanotechnology approaches for the treatment of solid tumors. Exp. Hematol. Oncol. 2025, 14, 76. [Google Scholar] [CrossRef]
  57. Da Silva, F.L.O.; Marques, M.B.F.; Kato, K.C.; Carneiro, G. Nanonization techniques to overcome poor water-solubility with drugs. Expert Opin. Drug Discov. 2020, 15, 853–864. [Google Scholar] [CrossRef]
  58. Cho, S.; Rasoulianboroujeni, M.; Kang, R.H.; Kwon, G.S. From Conventional to Next-Generation Strategies: Recent Advances in Polymeric Micelle Preparation for Drug Delivery. Pharmaceutics 2025, 17, 1360. [Google Scholar] [CrossRef]
  59. Nsairat, H.; Khater, D.; Sayed, U.; Odeh, F.; Al Bawab, A.; Alshaer, W. Liposomes: Structure, composition, types, and clinical applications. Heliyon 2022, 8, e09394. [Google Scholar] [CrossRef]
  60. Lin, S.; Tang, J.; Zhang, K.; Chen, Y.; Gao, R.; Yin, H.; Qin, L.C. Tuning oxygen-containing functional groups of graphene for supercapacitors with high stability. Nanoscale Adv. 2023, 5, 1163–1171. [Google Scholar] [CrossRef]
  61. Sivadasan, D.; Sultan, M.H.; Madkhali, O.; Almoshari, Y.; Thangavel, N. Polymeric Lipid Hybrid Nanoparticles (PLNs) as Emerging Drug Delivery Platform-A Comprehensive Review of Their Properties, Preparation Methods, and Therapeutic Applications. Pharmaceutics 2021, 13, 1291. [Google Scholar] [CrossRef]
  62. Chang, J.E.; Cho, H.J.; Yi, E.; Kim, D.D.; Jheon, S. Hypocrellin B and paclitaxel-encapsulated hyaluronic acid-ceramide nanoparticles for targeted photodynamic therapy in lung cancer. J. Photochem. Photobiol. B 2016, 158, 113–121. [Google Scholar] [CrossRef]
  63. Li, J.; Yao, S.; Wang, K.; Lu, Z.; Su, X.; Li, L.; Yuan, C.; Feng, J.; Yan, S.; Kong, B.; et al. Hypocrellin B-loaded, folate-conjugated polymeric micelle for intraperitoneal targeting of ovarian cancer in vitro and in vivo. Cancer Sci. 2018, 109, 1958–1969. [Google Scholar] [CrossRef]
  64. Li, T.; Hou, X.; Deng, H.; Zhao, J.; Huang, N.; Zeng, J.; Chen, H.; Gu, Y. Liposomal hypocrellin B as a potential photosensitizer for age-related macular degeneration: Pharmacokinetics, photodynamic efficacy, and skin phototoxicity in vivo. Photochem. Photobiol. Sci. 2015, 14, 972–981. [Google Scholar] [CrossRef]
  65. Zhou, L.; Jiang, H.; Wei, S.; Ge, X.; Zhou, J.; Shen, J. High-efficiency loading of hypocrellin B on graphene oxide for photodynamic therapy. Carbon 2012, 50, 5594–5604. [Google Scholar] [CrossRef]
  66. Zhang, Z.; Li, D.; Cao, Y.; Wang, Y.; Wang, F.; Zhang, F.; Zheng, S. Biodegradable Hypocrellin B nanoparticles coated with neutrophil membranes for hepatocellular carcinoma photodynamics therapy effectively via JUNB/ROS signaling. Int. Immunopharmacol. 2021, 99, 107624. [Google Scholar] [CrossRef] [PubMed]
  67. Bai, D.; Xia, X.; Yow, C.M.; Chu, E.S.; Xu, C. Hypocrellin B-encapsulated nanoparticle-mediated rev-caspase-3 gene transfection and photodynamic therapy on tumor cells. Eur. J. Pharmacol. 2011, 650, 496–500. [Google Scholar] [CrossRef] [PubMed]
  68. Li, Z.; Wang, J.; Chen, J.; Lei, W.; Wang, X.; Zhang, B. Hypocrellin B doped and pH-responsive silica nanoparticles for photodynamic therapy. Sci. China Chem. 2010, 53, 1994–1999. [Google Scholar] [CrossRef]
  69. Zheng, X.; Ge, J.; Wu, J.; Liu, W.; Guo, L.; Jia, Q.; Ding, Y.; Zhang, H.; Wang, P. Biodegradable hypocrellin derivative nanovesicle as a near-infrared light-driven theranostic for dually photoactive cancer imaging and therapy. Biomaterials 2018, 185, 133–141. [Google Scholar] [CrossRef] [PubMed]
  70. Wang, H.; Jia, Q.; Liu, W.; Nan, F.; Zheng, X.; Ding, Y.; Ren, H.; Wu, J.; Ge, J. Hypocrellin Derivative-Loaded Calcium Phosphate Nanorods as NIR Light-Triggered Phototheranostic Agents with Enhanced Tumor Accumulation for Cancer Therapy. ChemMedChem 2020, 15, 177–181. [Google Scholar] [CrossRef]
  71. Kang, W.; Zhao, F.; Cheng, J.; Feng, K.; Yan, L.; You, Y.; Li, J.; Meng, J. Comparative Study of Free and Encapsulated Hypocrellin B on Photophysical-Chemical Properties, Cellular Uptake, Subcellular Distribution, and Phototoxicity. Nanomaterials 2025, 15, 889. [Google Scholar] [CrossRef]
  72. Qi, S.; Guo, L.; Yan, S.; Lee, R.J.; Yu, S.; Chen, S. Hypocrellin A-based photodynamic action induces apoptosis in A549 cells through ROS-mediated mitochondrial signaling pathway. Acta Pharm. Sin. B 2019, 9, 279–293. [Google Scholar] [CrossRef]
  73. Zhang, J.; Cao, E.H.; Li, J.F.; Zhang, T.C.; Ma, W.J. Photodynamic effects of hypocrellin A on three human malignant cell lines by inducing apoptotic cell death. J. Photochem. Photobiol. B 1998, 43, 106–111. [Google Scholar] [CrossRef]
  74. Chio-Srichan, S.; Oudrhiri, N.; Bennaceur-Griscelli, A.; Turhan, A.G.; Dumas, P.; Refregiers, M. Toxicity and phototoxicity of Hypocrellin A on malignant human cell lines, evidence of a synergistic action of photodynamic therapy with Imatinib mesylate. J. Photochem. Photobiol. B 2010, 99, 100–104. [Google Scholar] [CrossRef]
  75. Li, R.; Yang, T.; Peng, X.; Feng, Q.; Hou, Y.; Zhu, J.; Chu, D.; Duan, X.; Zhang, Y.; Zhang, M. Enhancing the Photosensitivity of Hypocrellin A by Perylene Diimide Metallacage-Based Host-Guest Complexation for Photodynamic Therapy. Nanomicro Lett. 2024, 16, 226. [Google Scholar] [CrossRef]
  76. Jin, S.; Zhou, L.; Gu, Z.; Tian, G.; Yan, L.; Ren, W.; Yin, W.; Liu, X.; Zhang, X.; Hu, Z.; et al. A new near infrared photosensitizing nanoplatform containing blue-emitting up-conversion nanoparticles and hypocrellin A for photodynamic therapy of cancer cells. Nanoscale 2013, 5, 11910–11918. [Google Scholar] [CrossRef]
  77. Lin, X.; Yan, S.Z.; Qi, S.S.; Xu, Q.; Han, S.S.; Guo, L.Y.; Zhao, N.; Chen, S.L.; Yu, S.Q. Transferrin-Modified Nanoparticles for Photodynamic Therapy Enhance the Antitumor Efficacy of Hypocrellin A. Front. Pharmacol. 2017, 8, 815. [Google Scholar] [CrossRef]
  78. Khatoon, K.; Afthab, J.; Zhang, Z.; Chu, M.Q.; Huang, Y.; Li, J.; Wang, B.; Pu, G.; Zhou, C.H. Hypocrellin A-cisplatin-intercalated hectorite nano formulation for chemo-photodynamic tumor-targeted synergistic therapy. J. Mater. Sci. 2024, 5, 2087–2103. [Google Scholar] [CrossRef]
  79. Hopper, C. Photodynamic therapy: A clinical reality in the treatment of cancer. Lancet Oncol. 2000, 1, 212–219. [Google Scholar] [CrossRef]
  80. Kubrak, T.P.; Kołodziej, P.; Sawicki, J.; Mazur, A.; Koziorowska, K.; Aebisher, D. Some Natural Photosensitizers and Their Medicinal Properties for Use in Photodynamic Therapy. Molecules 2022, 27, 1192. [Google Scholar] [CrossRef]
  81. Tian, C.; Xu, S.; Chen, S.; Shen, J.; Zhang, M.; Shen, T. Chelation of hypocrellin B with zinc ions with electron paramagnetic resonance (EPR) evidence of the photodynamic activity of the resulting chelate. Free Radic. Res. 2001, 35, 543–554. [Google Scholar] [CrossRef] [PubMed]
  82. Lee, A.G. Lipid-protein interactions. Biochem. Soc. Trans. 2011, 39, 761–766. [Google Scholar] [CrossRef]
  83. Tiwari, G.; Tiwari, R.; Sriwastawa, B.; Bhati, L.; Pandey, S.; Pandey, P.; Bannerjee, S.K. Drug delivery systems: An updated review. Int. J. Pharm. Investig. 2012, 2, 2–11. [Google Scholar] [CrossRef]
  84. Kah, G.; Chandran, R.; Abrahamse, H. Curcumin a Natural Phenol and Its Therapeutic Role in Cancer and Photodynamic Therapy: A Review. Pharmaceutics 2023, 15, 639. [Google Scholar] [CrossRef]
  85. Lopes, T.Z.; de Moraes, F.R.; Tedesco, A.C.; Arni, R.K.; Rahal, P.; Calmon, M.F. Berberine associated photodynamic therapy promotes autophagy and apoptosis via ROS generation in renal carcinoma cells. Biomed. Pharmacother. 2020, 123, 109794. [Google Scholar] [CrossRef] [PubMed]
  86. Hu, J.; Song, J.; Tang, Z.; Wei, S.; Chen, L.; Zhou, R. Hypericin-mediated photodynamic therapy inhibits growth of colorectal cancer cells via inducing S phase cell cycle arrest and apoptosis. Eur. J. Pharmacol. 2021, 900, 174071. [Google Scholar] [CrossRef] [PubMed]
  87. Chen, Q.; Tian, S.; Zhu, J.; Li, K.T.; Yu, T.H.; Yu, L.H.; Bai, D.Q. Exploring a Novel Target Treatment on Breast Cancer: Aloe-emodin Mediated Photodynamic Therapy Induced Cell Apoptosis and Inhibited Cell Metastasis. Anti-Cancer Agents Med. Chem. 2016, 16, 763–770. [Google Scholar] [CrossRef]
  88. Yan, S.; Liao, X.; Xiao, Q.; Huang, Q.; Huang, X. Photostabilities and anti-tumor effects of curcumin and curcumin-loaded polydopamine nanoparticles. RSC Adv. 2024, 14, 13694–13702. [Google Scholar] [CrossRef]
  89. Marques, C.; Fernandes, M.H.; Lima, S.A.C. Elucidating Berberine’s Therapeutic and Photosensitizer Potential Through Nanomedicine Tools. Pharmaceutics 2023, 15, 2282. [Google Scholar] [CrossRef]
  90. Kubin, A.; Loew, H.G.; Burner, U.; Jessner, G.; Kolbabek, H.; Wierrani, F. How to make hypericin water-soluble. Pharmazie 2008, 63, 263–269. [Google Scholar]
  91. Narayanan, S.; Jadhav, A.P.; Kadam, V.J. Forced Degradation Studies of Aloe Emodin and Emodin by HPTLC. Indian J. Pharm. Sci. 2015, 77, 795–798. [Google Scholar]
  92. Marquardt, J.U.; Gomez-Quiroz, L.; Arreguin Camacho, L.O.; Pinna, F.; Lee, Y.H.; Kitade, M.; Domínguez, M.P.; Castven, D.; Breuhahn, K.; Conner, E.A.; et al. Curcumin effectively inhibits oncogenic NF-κB signaling and restrains stemness features in liver cancer. J. Hepatol. 2015, 63, 661–669. [Google Scholar] [CrossRef]
  93. Tian, B.; Zhao, Y.; Liang, T.; Ye, X.; Li, Z.; Yan, D.; Fu, Q.; Li, Y. Curcumin inhibits urothelial tumor development by suppressing IGF2 and IGF2-mediated PI3K/AKT/mTOR signaling pathway. J. Drug Target. 2017, 25, 626–636. [Google Scholar] [CrossRef] [PubMed]
  94. Talib, W.H.; Al-Hadid, S.A.; Ali, M.B.W.; Al-Yasari, I.H.; Ali, M.R.A. Role of curcumin in regulating p53 in breast cancer: An overview of the mechanism of action. Breast Cancer (Dove Med. Press). 2018, 10, 207–217. [Google Scholar] [CrossRef]
  95. Kim, H.S.; Kim, M.J.; Kim, E.J.; Yang, Y.; Lee, M.S.; Lim, J.S. Berberine-induced AMPK activation inhibits the metastatic potential of melanoma cells via reduction of ERK activity and COX-2 protein expression. Biochem. Pharmacol. 2012, 83, 385–394. [Google Scholar] [CrossRef]
  96. Kim, S.A.; Kwon, Y.; Kim, J.H.; Muller, M.T.; Chung, I.K. Induction of topoisomerase II-mediated DNA cleavage by a protoberberine alkaloid, berberrubine. Biochemistry 1998, 37, 16316–16324. [Google Scholar] [CrossRef]
  97. Tsang, C.M.; Cheung, Y.C.; Lui, V.W.; Yip, Y.L.; Zhang, G.; Lin, V.W.; Cheung, K.C.; Feng, Y.; Tsao, S.W. Berberine suppresses tumorigenicity and growth of nasopharyngeal carcinoma cells by inhibiting STAT3 activation induced by tumor associated fibroblasts. BMC Cancer 2013, 13, 619. [Google Scholar] [CrossRef]
  98. Huntosova, V.; Stroffekova, K. Hypericin in the Dark: Foe or Ally in Photodynamic Therapy? Cancers 2016, 8, 93. [Google Scholar] [CrossRef]
  99. Van de Putte, M.; Roskams, T.; Vandenheede, J.R.; Agostinis, P.; de Witte, P.A. Elucidation of the tumoritropic principle of hypericin. Br. J. Cancer 2005, 92, 1406–1413. [Google Scholar] [CrossRef]
  100. Ali, S.M.; Olivo, M. Bio-distribution and subcellular localization of Hypericin and its role in PDT induced apoptosis in cancer cells. Int. J. Oncol. 2002, 21, 531–540. [Google Scholar] [CrossRef]
  101. Yang, X.; Chen, W.; Sun, H.; Chen, W.; Xu, W.; He, C.; Liu, Y.; Kuang, Y.; Ma, Y.; Zhong, B.; et al. Emodin promotes GSK-3β-mediated PD-L1 proteasomal degradation and enhances anti-tumor immunity in hepatocellular carcinoma. Chin. Med. 2025, 20, 126. [Google Scholar] [CrossRef]
  102. Zhang, Y.; Pu, W.; Bousquenaud, M.; Cattin, S.; Zaric, J.; Sun, L.K.; Rüegg, C. Emodin Inhibits Inflammation, Carcinogenesis, and Cancer Progression in the AOM/DSS Model of Colitis-Associated Intestinal Tumorigenesis. Front. Oncol. 2021, 10, 564674. [Google Scholar] [CrossRef]
  103. Ma, J.; Lu, H.; Wang, S.; Chen, B.; Liu, Z.; Ke, X.; Liu, T.; Fu, J. The anthraquinone derivative Emodin inhibits angiogenesis and metastasis through downregulating Runx2 activity in breast cancer. Int. J. Oncol. 2015, 46, 1619–1628. [Google Scholar] [CrossRef]
  104. Henriques, J.; Pina, J.; Braga, M.E.M.; Dias, A.M.A.; Coimbra, P.; de Sousa, H.C. Novel Oxygen- and Curcumin-Laden Ionic Liquid@Silica Nanocapsules for Enhanced Antimicrobial Photodynamic Therapy. Pharmaceutics 2023, 15, 1080. [Google Scholar] [CrossRef]
  105. Hirakawa, K.; Hirano, T. The microenvironment of DNA switches the activity of singlet oxygen generation photosensitized by berberine and palmatine. Photochem. Photobiol. 2008, 84, 202–208. [Google Scholar] [CrossRef]
  106. Barras, A.; Boussekey, L.; Courtade, E.; Boukherroub, R. Hypericin-loaded lipid nanocapsules for photodynamic cancer therapy in vitro. Nanoscale 2013, 5, 10562–10572. [Google Scholar] [CrossRef]
  107. Liu, Y.; Zhao, J.; Xu, X.; Xu, Y.; Cui, W.; Yang, Y.; Li, J. Emodin-Based Nanoarchitectonics with Giant Two-Photon Absorption for Enhanced Photodynamic Therapy. Angew. Chem. Int. Ed. Engl. 2023, 62, e202308019. [Google Scholar] [CrossRef]
  108. Liu, Y.; Yin, R.; Tian, Y.; Xu, S.; Meng, X. Curcumin nanopreparations: Recent advance in preparation and application. Biomed. Mater. 2024, 19, 052009. [Google Scholar] [CrossRef]
  109. Fu, L.; Mou, J.; Deng, Y.; Ren, X. Structural modifications of berberine and their binding effects towards polymorphic deoxyribonucleic acid structures: A review. Front. Pharmacol. 2022, 13, 940282. [Google Scholar] [CrossRef]
  110. Lou, H.Y.; Li, Y.N.; Yi, P.; Jian, J.Y.; Hu, Z.X.; Gu, W.; Huang, L.J.; Li, Y.M.; Yuan, C.M.; Hao, X.J. Hyperfols A and B: Two Highly Modified Polycyclic Polyprenylated Acylphloroglucinols from Hypericum perforatum. Org. Lett. 2020, 22, 6903–6906. [Google Scholar] [CrossRef]
  111. Liang, X.Y.; Battini, N.; Sui, Y.F.; Ansari, M.F.; Gan, L.L.; Zhou, C.H. Aloe-emodin derived azoles as a new structural type of potential antibacterial agents: Design, synthesis, and evaluation of the action on membrane, DNA, and MRSA DNA isomerase. RSC Med. Chem. 2021, 12, 602–608. [Google Scholar] [CrossRef]
  112. Aggarwal, B.B.; Sundaram, C.; Malani, N.; Ichikawa, H. Curcumin: The Indian solid gold. Adv. Exp. Med. Biol. 2007, 595, 1–75. [Google Scholar]
  113. Neag, M.A.; Mocan, A.; Echeverría, J.; Pop, R.M.; Bocsan, C.I.; Crişan, G.; Buzoianu, A.D. Berberine: Botanical Occurrence, Traditional Uses, Extraction Methods, and Relevance in Cardiovascular, Metabolic, Hepatic, and Renal Disorders. Front. Pharmacol. 2018, 9, 557. [Google Scholar] [CrossRef]
  114. Peng, Z.; Lu, J.; Liu, K.; Xie, L.; Wang, Y.; Cai, C.; Yang, D.; Xi, J.; Yan, C.; Li, X.; et al. Hypericin as a promising natural bioactive naphthodianthrone: A review of its pharmacology, pharmacokinetics, toxicity, and safety. Phytother. Res. 2023, 37, 5639–5656. [Google Scholar] [CrossRef]
  115. Zheng, Q.; Li, S.; Li, X.; Liu, R. Advances in the study of emodin: An update on pharmacological properties and mechanistic basis. Chin. Med. 2021, 16, 102. [Google Scholar] [CrossRef]
  116. Wang, X.; Peng, J.; Meng, C.; Feng, F. Recent advances for enhanced photodynamic therapy: From new mechanisms to innovative strategies. Chem. Sci. 2024, 15, 12234–12257. [Google Scholar] [CrossRef]
  117. Zeeshan, T. Curcumin VS Photo-Bio-Modulation Therapy of Oral Mucositis in Pediatric Patients Undergoing Anti-Cancer Non-invasive Treatment. 2024. Available online: https://clinicaltrials.gov/study/NCT06044142 (accessed on 31 October 2025).
  118. Dryden, G.W. Study Investigating the Ability of Plant Exosomes to Deliver Curcumin to Normal and Colon Cancer Tissue. 2023. Available online: https://clinicaltrials.gov/study/NCT01294072 (accessed on 17 November 2025).
  119. Morrow, G. Prophylactic Topical Agents in Reducing Radiation-Induced Dermatitis in Patients with Non-Inflammatory Breast Cancer (Curcumin-II). 2017. Available online: https://www.clinicaltrials.gov/study/NCT02556632 (accessed on 17 November 2025).
  120. Baskaran, R.; Lee, J.; Yang, S.G. Clinical development of photodynamic agents and therapeutic applications. Biomater. Res. 2018, 22, 25. [Google Scholar] [CrossRef] [PubMed]
  121. Kim, T.E.; Chang, J.E. Recent Studies in Photodynamic Therapy for Cancer Treatment: From Basic Research to Clinical Trials. Pharmaceutics 2023, 15, 2257. [Google Scholar] [CrossRef]
  122. Crusca, J.d.S.; de Moraes, L.H.O.; Figueira, T.G.; Parizotto, N.A.; Rodrigues, G.J. Photodynamic Therapy Effects with Curcuma longa L. Active Ingredients in Gel and Blue LED on Acne: A Randomized, Controlled, and Double-Blind Clinical Study. Photonics 2025, 12, 80. [Google Scholar] [CrossRef]
  123. Allison, R.R.; Ferguson, J.S. Photodynamic therapy to a primary cancer of the peripheral lung: Case report. Photodiagnosis Photodyn. Ther. 2022, 39, 103001. [Google Scholar] [CrossRef]
  124. Gold, M.H. Therapeutic and aesthetic uses of photodynamic therapy part one of a five-part series: The use of photodynamic therapy in the treatment of actinic keratoses and in photorejuvenation. J. Clin. Aesthet. Dermatol. 2008, 1, 32–37. [Google Scholar]
  125. Law, S.K.; Leung, A.W.N.; Xu, C. Photodynamic Action of Curcumin and Methylene Blue Against Bacteria and SARS-CoV-2—A Review. Pharmaceuticals 2024, 17, 34. [Google Scholar] [CrossRef]
  126. Penetra, M.; Arnaut, L.G.; Gomes-da-Silva, L.C. Trial watch: An update of clinical advances in photodynamic therapy and its immunoadjuvant properties for cancer treatment. Oncoimmunology 2023, 12, 2226535. [Google Scholar] [CrossRef]
  127. Zhao, Y.Y.; Lu, L.; Jeong, H.; Kim, H.; Li, X.; Zhang, H.; Yoon, J. Enhancing biosafety in photodynamic therapy: Progress and perspectives. Chem. Soc. Rev. 2025, 54, 7749–7768. [Google Scholar] [CrossRef]
  128. Grin, M.; Suvorov, N.; Ostroverkhov, P.; Pogorilyy, V.; Kirin, N.; Popov, A.; Sazonova, A.; Filonenko, E. Advantages of combined photodynamic therapy in the treatment of oncological diseases. Biophys. Rev. 2022, 14, 941–963. [Google Scholar] [CrossRef]
  129. Zhang, S.; Chen, W.; Zhou, J.; Liang, Q.; Zhang, Y.; Su, M.; Zhang, Z.; Qu, J. The Benefits and Safety of Monoclonal Antibodies: Implications for Cancer Immunotherapy. J. Inflamm. Res. 2025, 18, 4335–4357. [Google Scholar] [CrossRef]
  130. Peng, X.; Liu, Z.; Luo, C.; Sun, R.; Zhang, Y.; Li, B.; Zou, Y.; Zhu, J.; Yuan, R. PSMD12 promotes hepatocellular carcinoma progression by stabilizing CDK1. Front. Immunol. 2025, 16, 1581398. [Google Scholar] [CrossRef]
  131. Wang, Z.B.; Zhang, X.; Fang, C.; Liu, X.T.; Liao, Q.J.; Wu, N.; Wang, J. Immunotherapy and the ovarian cancer microenvironment: Exploring potential strategies for enhanced treatment efficacy. Immunology 2024, 173, 14–32. [Google Scholar] [CrossRef]
Figure 1. Chemical structure of (a) Hypocrellin A (HA) and (b) Hypocrellin B (HB).
Figure 1. Chemical structure of (a) Hypocrellin A (HA) and (b) Hypocrellin B (HB).
Pharmaceuticals 18 01847 g001
Figure 2. Absorption (solid line) and fluorescence (dashed line) of (a) Hypocrellin A (HA); absorption (triangle with line) and fluorescence (spot with line) of (b) Hypocrellin B (HB) in DMSO. Copied from references [40,41].
Figure 2. Absorption (solid line) and fluorescence (dashed line) of (a) Hypocrellin A (HA); absorption (triangle with line) and fluorescence (spot with line) of (b) Hypocrellin B (HB) in DMSO. Copied from references [40,41].
Pharmaceuticals 18 01847 g002
Table 3. Examples of the traditional strategy of HA-PDT for cancer.
Table 3. Examples of the traditional strategy of HA-PDT for cancer.
StudyExperiment ParametersConsequenceReferences
1Hypocrellin A-based photodynamic action induces apoptosis in A549 cells through ROS-mediated mitochondrial signaling pathwayAbsorption (λabs): 540 to 560 nm (dominant band), secondary peak at 465 nm.
Absorption maximum (λabs max): 550 nm for molar extinction coefficients (ε): ~3.1 × 104 M−1·cm−1
Emission (λem): 630 to 660 nm with λem max: 645 nm
Singlet oxygen quantum yields (ΦΔ): ~0.63
Cytotoxicity IC50: ~0.8 to 1.2 μM in A549 cells
HA-PDT was associated with cell shrinkage, externalization of cell membrane phosphatidylserine, DNA fragmentation, and mitochondrial disruption, as well as pronounced release of cytochrome c, and activation of caspase-3, -9, and -7.[72]
2Photodynamic effects of hypocrellin A on three human malignant cell lines by inducing apoptotic cell deathAbsorption (λabs): 540 to 560 nm (dominant band), secondary peak at 465 nm.
Absorption maximum (λabs max): 550 nm for molar extinction coefficients (ε): ~3.0 × 104 M−1·cm−1
Emission (λem): 630 to 660 nm with λem max: 645 nm
Singlet oxygen quantum yields (ΦΔ): ~0.63
Cytotoxicity IC50: ~0.8 to 1.5 μM in HeLa, MGC-803, and HIC cells
HA-PDT induced apoptosis or necrosis, evidenced by morphological changes, DNA fragmentation, and a decrease in mitochondrial dehydrogenase activity in human malignant epithelioid cells.[73]
3Toxicity and phototoxicity of hypocrellin A on malignant human cell lines, evidence of a synergistic action of photodynamic therapy with Imatinib mesylateAbsorption (λabs): 540 to 560 nm (dominant band), secondary peak at 465 nm.
Absorption maximum (λabs max): 550 nm for molar extinction coefficients (ε): ~3.0 × 104 M−1·cm−1
Emission (λem): 630 to 660 nm with λem max: 645 nm
Singlet oxygen quantum yields (ΦΔ): ~0.63
Cytotoxicity IC50: ~0.6 to 1.5 μM in HeLa, Calu, and K562 cell lines
The phototoxicity of HA in epithelial cell lines demonstrated a synergy between imatinib mesylate and photodynamic therapy to circumvent imatinib mesylate resistance.[74]
Table 4. Examples of the nanotechnology of HA-PDT for cancer.
Table 4. Examples of the nanotechnology of HA-PDT for cancer.
StudyExperiment ParametersConsequenceReferences
1Enhancing the photosensitivity of hypocrellin A by perylene diimide metallacage-based host-guest complexation for photodynamic therapyAbsorption (λabs): 540 to 560 nm (dominant band), secondary peak at 465 nm.
Absorption maximum (λabs max): 550 nm for molar extinction coefficients (ε): ~3.0 to 3.6 × 104 M−1·cm−1
Emission (λem): 630 to 660 nm with λem max: 645 nm
Singlet oxygen quantum yields (ΦΔ): ~0.63 to 0.72
Cytotoxicity IC50: ~0.5 to 1.2 μM in HeLa, MCF-7 cells
In vivo pharmacokinetics: reduce systemic toxicity
HA perylene diimide-based metallacages displayed excellent anticancer activities upon light irradiation in PDT and enhanced the photosensitivity of conventional photosensitizers via host-guest complexation-based fluorescence resonance energy transfer.[75]
2A new near-infrared photosensitizing nanoplatform containing blue-emitting up-conversion nanoparticles and hypocrellin A for photodynamic therapy of cancer cellsAbsorption (λabs): 540 to 560 nm (dominant band), secondary peak at 465 nm.
Absorption maximum (λabs max): 550 nm for molar extinction coefficients (ε): ~3.0 × 104 M−1·cm−1
Emission (λem): 630 to 660 nm with λem max: 645 nm
Singlet oxygen quantum yields (ΦΔ): ~0.65
Cytotoxicity IC50: ~0.6 to 1.0 μM in HeLa, and MCF-7 cells
Tween 20-up-conversion nanoparticles@HA complexes-PDT efficiently produced singlet oxygen to kill cancer cells, exhibited positive contrast effects on the magnetic resonance imaging (MRI) and computed tomography (CT) imaging.[76]
3Transferrin-modified nanoparticles for photodynamic therapy enhance the antitumor efficacy of Hypocrellin AAbsorption (λabs): 540 to 560 nm (dominant band), secondary peak at 465 nm.
Absorption maximum (λabs max): 550 nm for molar extinction coefficients (ε): ~3.0 × 104 M−1·cm−1
Emission (λem): 630 to 660 nm with λem max: 645 nm
Singlet oxygen quantum yields (ΦΔ): ~0.63
Cytotoxicity IC50: ~0.6 to 0.9 μM in HeLa, and HepG2 cells
Poly(D, L-Lactide-co-glycolide) and carboxymethyl chitosan nanoparticle-loaded with HA enhanced PDT therapeutic efficacy, which caused cell apoptosis in tumor tissue and slight side effects in normal organs. [77]
4Hypocrellin A-cisplatin-intercalated hectorite nano formulation for chemo-photodynamic tumor-targeted synergistic therapyAbsorption (λabs): 540 to 560 nm (dominant band), secondary peak at 465 nm.
Absorption maximum (λabs max): 550 nm for molar extinction coefficients (ε): ~3.0 × 104 M−1·cm−1
Emission (λem): 630 to 660 nm with λem max: 645 nm
Singlet oxygen quantum yields (ΦΔ): ~0.62
Cytotoxicity IC50: ~0.5 to 0.8 μM in synergistic therapy with cisplatin
HA-cisplatin-intercalated hectorite nano formulation-PDT possessed stable light absorption, high oxygen generation with controlled drug release efficacy to induce apoptosis and necrosis for targeted and effective esophageal cancer treatment. [78]
Table 5. A comparative study of HA and HB in PDT.
Table 5. A comparative study of HA and HB in PDT.
Hypocrellin B (HB)Hypocrellin A (HA)
Absorption and emission peaksAbsorption (λabs): 540 to 560 nm (dominant band), secondary peak at 465 nm,
Absorption maximum (λabs max): 550 nm;
Emission (λem): 630 to 660 nm
with λem max: 645 nm
Absorption (λabs): 540 to 560 nm (dominant band), secondary peak at 465 nm,
Absorption maximum (λabs max): 550 nm;
Emission (λem): 630 to 660 nm
with λem max: 645 nm
Single oxygen yieldHigh (0.50 or above, depend on the solvent)High (0.60 or above, depend on the solvent)
In vitro studiesInvestigate different cancer cell lines (Table 1)Slightly limited than HB (Table 3)
In vivo studiesMice modelsSlightly limited than HB
PharmacokineticsSelective target and uptake for cancerSimilarly to HB
Toxicity profileLow dark toxicityLow dark toxicity
Table 6. Benefits of using HA or HB over the other TCM PSs.
Table 6. Benefits of using HA or HB over the other TCM PSs.
PSSource Photo-StabilityTumor SelectivitySinglet Oxygen YieldStructural Modification
Hypocrellin
(HA and HB)
Fungus Hypocrella bambusaeExcellentExcellent
(After chemical and structural modification)
HighHigh
CurcuminCurcuma longa [112]PoorSatisfactionLowLimit
BerberineCoptidis rhizome [113]SatisfactionSatisfactionAverageLimit
HypericinSt. John’s wort [114]GoodGoodHighSatisfaction
EmodinRhubarb [115]SatisfactionSatisfactionHighSatisfaction
Table 7. Clinical stage and regulatory status of synthetic vs. natural PSs.
Table 7. Clinical stage and regulatory status of synthetic vs. natural PSs.
Synthetic PSs (Photofrin II, Methyl Aminolevulinate)Natural PSs (Curcumin, HB)
Clinical stageCompleted the multiple indications for cancers [120,121]Curcumin: clinical phase II [122]
HB: Not in clinical phase and still in pre-clinical, in vitro, and in vivo studies
Regulatory statusApproved by the Food and Drug Administration (FDA) [123,124]Curcumin and HB are not FDA-approved [55,125]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Huang, J.; Law, S.K.; Leung, A.W.N.; Xu, C. Photodynamic Action of Hypocrellin A and Hypocrellin B Against Cancer—A Review. Pharmaceuticals 2025, 18, 1847. https://doi.org/10.3390/ph18121847

AMA Style

Huang J, Law SK, Leung AWN, Xu C. Photodynamic Action of Hypocrellin A and Hypocrellin B Against Cancer—A Review. Pharmaceuticals. 2025; 18(12):1847. https://doi.org/10.3390/ph18121847

Chicago/Turabian Style

Huang, Jinju, Siu Kan Law, Albert Wing Nang Leung, and Chuanshan Xu. 2025. "Photodynamic Action of Hypocrellin A and Hypocrellin B Against Cancer—A Review" Pharmaceuticals 18, no. 12: 1847. https://doi.org/10.3390/ph18121847

APA Style

Huang, J., Law, S. K., Leung, A. W. N., & Xu, C. (2025). Photodynamic Action of Hypocrellin A and Hypocrellin B Against Cancer—A Review. Pharmaceuticals, 18(12), 1847. https://doi.org/10.3390/ph18121847

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Article metric data becomes available approximately 24 hours after publication online.
Back to TopTop